Parkinson's & Alzheimer's Flashcards

1
Q

What is Parkinson’s caused by? briefly

What are the 2 ways centrally acting drugs in the CNS can act?

Why is it more difficult to target drugs at the CNS?

What 3 things need to be considered with CNS drugs?

What are the 3 most common issues?

A

Loss of dopaminergic substantia nigra cells that produce dopamine

  1. Increase receptor signalling by blocking uptake, supplying precursor or by direct activation (anxiety & benzodiazepines)
  2. Decrease transmitter function as antagonist

Very hard to measure/quantify symptoms (e.g Schizophrenia) and so many neurones & contacts

  1. Multiple receptors - e.g 5HT has several excitatory & inhibitory
  2. Receptors can up & down regulate drugs
  3. CNS is so complex - many synaptic connections & transmitters can act on different sites in brain

kk

  1. Side effects (with many transmitters acting different receptors in brain & sites)
  2. Reaching the right target - metabolism, blood brain barrier BBB (lipid soluble, ionisation)
  3. Compensation (trying to offset drug) - by altered receptor expression (reduction with opioids so have to increase amount drug) and general adaptation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How would you solve a drug that can’t penetrate the BBB?

How would you solve reduced drug metabolism?

Side effects?

Drug acting at unwanted sites?

Compensation?

A

Disrupting the BBB by intra-arterial mannitol infusion & focused ultrasound

Adjunct given - drug given with the drug to increase its effect - helping drug reach the CNS e.g blocks enzymes being broken down in periphery

Use adjunct antagonist blocking receptors which don’t want to activate

Receptors in periphery & brain - use antagonist adjunct that doesn’t pass BBB & blocks peripheral receptors

Increased drug dosage e.g with opiates OR if been on for long time can gradually reduce dose so system acclimatises so don’t pull off immediately

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the early characterisations of Parkinson’s? 4

What are the later characteristics of Parkinson’s disease? 7

What is Parkinson’s caused by?

A
  1. Resting pin-rolling tremor
  2. Bradykinesia (slowing of movement)
  3. Anosmia
  4. Postural hypotension - drop blood pressure on standing

kk

  1. Akinesia (difficulty initiating movement)
  2. Stone-faced/mask-like face - no facial muscle movement
  3. Micrographia (smaller handwriting)
  4. Hypophonia (decreased volume of speaking) & monotonous speach
  5. Rigidity in joints - Parkinson’s shuffle
  6. Postural instability causing anteropulsion (stooped posture)
  7. Memory deficits - Lewy bodies

Neurodegeneration of substantia nigra (production of dopamine) - synuclein & tau accumulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the basal ganglia made up of?

What is the globus pallidus made up of?

What is the substantial nigra made up of?

What is the striatum in the rat/mouse equivalent to in humans?

What is the striatum’s role?

What does the basal ganglia in the brain modulate?

A

Caudate, lentiform nucleus (putamen & globus pallidus), subthalamic nucleus (STN) and substantia nigra (SN)

Internal & external section

Pars compacta (dopaminergic) and reticulata (output of basal ganglia)

Caudate & putamen

Input of the basal ganglia (receives an output)

Movement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What are the steps of the direct (Go) pathway for movement? 3 steps

What are the steps of the indirect (No Go) pathway for movement? 4 steps

What is the difference between the two pathways?

What is the hyperdirect pathway?

What do these pathways collectively do?

A
  1. Motor commands received by the striatum from the cortex
  2. Striatum sends inhibitory signals to the substantia nigra reticulata & globus pallidus interior via the direct pathway
  3. The SNr & GPi send inhibitory signals to motor output areas, causing excitatory response of motor activity (2 x inhib = excitatory)

kk

  1. Motor commands received by the striatum from the cortex
  2. Striatum sends inhibitory signals to the globus pallidus external segment
  3. This sends inhibitory signals to subthalamic nucleus (STN) causing excitation of SNr/GPi
  4. This increases the SNr/GPi activity of its output and hence increased inhibition of motor output areas

Go = causes movement
No go = inhibits movement

Cortex sends excitatory signals directly to STN which sends excitatory signals directly to SNr/GPi causing increased activity & inhibition of motor output

Produce a balance between inhibitory & excitatory of motor output areas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What part of the substantial nigra is responsible for dopaminergic neurones & dopamine release?

When is dopamine released in the striatum?

What was the first experiment showing this correlation?

How did they show that this correlation was directly causing dopamine release?

What does dopamine then do? conclusion

A

Pars compacta

Association with movement

Injected virus into SN to selectively infect dopamingeric neurones & introduce GFP-calcium dependent molecule GCaMP6f - GFP calcium indicator such that on dopamine release (as a result of calcium voltage gates opening & calcium entering neurone in action potential causing release of dopamine) when mouse running on treadmill with fluorescence spectroscopy

Gene coding for channel rhodopsin 2 with YFP into dopaminergic cells such that when light was shone into the mouse, dopamine was released & mouse started running

Release of dopamine from substanial nigra compacta promotes motor activity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What does dopamine do from the substantial nigra compacta to initiate movement? 2 things

What is the problem in Parkinson’s?

How is the tremor caused?

A
  1. Excites direct pathway via D1 excitatory receptors (to inhibit SNr/GPi)
  2. Inhibits indirect no Go pathway via D2 inhibitory receptors

80% dopaminergic neurones in SNc are lost leading to less activation of Go pathway & less inhibition of indirect no go pathway

Reduced D2-mediated inhibition of striatal cholinergic neurones (over-excitation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the approaches to treating Parkinson’s? 4

Why is L-DOPA given & not dopamine?

What is the good thing of using L-DOPA?

What other pathways are activated too? 5 & their side effects

What is another issue of DA?

A
  1. Dopamine precursors (L-dopa) to increase dopamine synthesis
  2. Block reuptake of DA by blocking Dopamine Transporters DAT
  3. Inhibit enzymes in degradation - monoamine oxidase & catechol-o-methyltransferase
  4. Dopaminergic agonists that act on receptors

Dopamine will not cross the BBB

Increased movement due to increased activation of nigrostriatal pathway

  1. Mesolimbic - addictive/wanting/liking
  2. Mesocortical - psychotic effects
  3. Pituitary - hormonal effects - growth hormone release etc
  4. Chemoreceptor trigger zone (thin BBB to sense toxins) activated by D2 receptors - vomiting
  5. Periphery - drop in blood pressure by increased DA

Increased DA increased metabolism of noradrenaline by dopamine ß-hydroxylase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

How then does DA caused decreased blood pressure?

How is L-dopa produced?

How is L-dopa converted into Dopamine?

How is dopamine converted in the brain? 2 things

What are the 2 problems with giving L-DOPA (peripherally)?

How is this overcome?

How could you increase the amount of dopamine available in the brain?

A

Dopamine ß-hydroxylase cannot convert DA to NA quickly enough - so drop in noradrenergic signalling & hence lower blood pressure

By tyrosine with tyrosine hydroxylase

DOPA decarboxylase past the BBB

  1. mono-amineoxidase into DOPAC
  2. COMT into 3-MT

kk

  1. Side effects of dopamine in periphery
  2. Less L-DOPA available to cross BBB to brain

Adjunct therapy - carbidopa or benserazide - block DOPA decarboxylase (conversion of L-DOPA to dopamine in periphery) but not in the brain - allows L-DOPA in brain so fewer side effects caused in periphery

Selegiline = MAO inhibitor
Entacapone = COMT inhibitor - blocking enzymatic break down of dopamine with both to increase availability in presynapse & cleft

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

How is the chemoreceptor trigger zone affected?

How is vomiting induced?

How can you prevent activation of the chemoreceptor trigger zone in periphery?

Why is a different adjunct also used and what is it?

What is a good thing about this adjunct?

A

Dopamine produced in the periphery & the brain - as along the BBB

Activation of D2 receptors by dopamine

Carbidopa/benserazide to block DOPA decarboxylase (L-DOPA -> Dopamine)

Insufficient alone - use domperidone D2 receptor antagonist - in periphery & CTZ

Does not cross the BBB so can’t access other D2 receptors in CNS like striatum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Why is upregulating DA no longer effective in late parkinson’s?

What treatments are used in late Parkinson’s? 3

What other drug is given to prevent vomiting?

What is the main side effect of the treatments?

What other treatment can help treat Parkinson’s?

A

Lose dopaminergic neurones - only effective if have neurones to produce dopamine & release it into striatum

  1. Bromocriptine - D2 agonist
  2. Apomorphine - D2 & D1 agonist
  3. Amantadine - D2 & D1 agonist & others (nicotinic, NMDA receptors)

Domperidone - peripheral D2 antagonist as an adjunct

Increased impulsivity - gambling

DBS - deep brain stimulation - electrodes & electrical stimulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

How is Alzheimer’s disease characterised?

Which neurones are most vulnerable?

What are the symptoms of pre-dementia?

Early dementia?

Advanced?

A

Amyloid Aß plaques and tau (neurofibrillary) tangles causing extensive cell death and brain shrinkage

Acetylcholinergic

Short term memory loss & decline in executive function

More decline in short term memory & executive function, and some motor deficits

Long term memory impaired, procedural memories affected (how to ride a bike)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

How do amyloid plaques form?

How is the protein treated in healthy brains?

Alzheimer’s brains?

What are NFTs? (neurofibrillary tangles)

Where do they accumulate in early & late stages?

What main cells does Alzheimer’s target?

A

Accumulation of amyloid protein (ß-amyloid or Aß-amyloid) between neurones in the brain

Amyloid precursor protein APP (which produces amyloid) is cleaved by ß-secretase BACE1 & the product APP-CTFß is cleaved by gamma-secretase - APP protein fragments are degraded after use

APP protein fragments (Aß42) accumulate and can be surrounded by neutrites to form neutritic plaques or just dense amyloid deposits without neutrites (as they have died off - long term)

Fibres in cytoplasm of neurones made up of abnormally phosphorylated tau protein and ubiquitin

Entorhinal cortex in early stages & progresses to hippocampus & neocortex

Cholinergic neurones/cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is the main treatment of Alzheimer’s?

What is the effect?

What is another treatment?

What treatment targets the amyloid plaques?

What does spin-labelled fluorene compounds aim to do?

Why is antibiotics being looked at?

A

Acetylcholinesterase inhibitors - preventing the breakdown of acetylcholine to increase its presence in synapses - Donepezil, Rivastigmine, Galantamine

Not much - only slows cognitive decline but still achieved - many side effects

Memantine - NMDA receptor inhibitor as damaged neurones release significant amounts of glutamate - prevents further excitotoxic neurone damage through increased cell firing

Aducanuab - anti-amyloid antibody - which also has limited efficacy & side effects like the ones above

Reduce amyloid plaque formation in cultured neurones & cross the blood-brain barrier but only in early stage development

Relationship between gut microbiome & neuroinflammation - can decrease glutamate transport & neurone damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the side effects of AChE inhibitors?

What is the overall clinical effect of Donepezil? (10mg, 5mg, placebo)

A

Fatigue, insomnia, hallucinations, muscle cramps

Clinical improvement - leading to a slower cognitive decline

How well did you know this?
1
Not at all
2
3
4
5
Perfectly